Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Stem Cell Res Ther ; 15(1): 11, 2024 01 08.
Article in English | MEDLINE | ID: mdl-38185673

ABSTRACT

BACKGROUND: Mesenchymal stem cells in the adult corneal stroma (named corneal stromal stem cells, CSSCs) inhibit corneal inflammation and scarring and restore corneal clarity in pre-clinical corneal injury models. This cell therapy could alleviate the heavy reliance on donor materials for corneal transplantation to treat corneal opacities. Herein, we established Good Manufacturing Practice (GMP) protocols for CSSC isolation, propagation, and cryostorage, and developed in vitro quality control (QC) metric for in vivo anti-scarring potency of CSSCs in treating corneal opacities. METHODS: A total of 24 donor corneal rims with informed consent were used-18 were processed for the GMP optimization of CSSC culture and QC assay development, while CSSCs from the remaining 6 were raised under GMP-optimized conditions and used for QC validation. The cell viability, growth, substrate adhesion, stem cell phenotypes, and differentiation into stromal keratocytes were assayed by monitoring the electric impedance changes using xCELLigence real-time cell analyzer, quantitative PCR, and immunofluorescence. CSSC's conditioned media were tested for the anti-inflammatory activity using an osteoclastogenesis assay with mouse macrophage RAW264.7 cells. In vivo scar inhibitory outcomes were verified using a mouse model of anterior stromal injury caused by mechanical ablation using an Algerbrush burring. RESULTS: By comparatively assessing various GMP-compliant reagents with the corresponding non-GMP research-grade chemicals used in the laboratory-based protocols, we finalized GMP protocols covering donor limbal stromal tissue processing, enzymatic digestion, primary CSSC culture, and cryopreservation. In establishing the in vitro QC metric, two parameters-stemness stability of ABCG2 and nestin and anti-inflammatory ability (rate of inflammation)-were factored into a novel formula to calculate a Scarring Index (SI) for each CSSC batch. Correlating with the in vivo scar inhibitory outcomes, the CSSC batches with SI < 10 had a predicted 50% scar reduction potency, whereas cells with SI > 10 were ineffective to inhibit scarring. CONCLUSIONS: We established a full GMP-compliant protocol for donor CSSC cultivation, which is essential toward clinical-grade cell manufacturing. A novel in vitro QC-in vivo potency correlation was developed to predict the anti-scarring efficacy of donor CSSCs in treating corneal opacities. This method is applicable to other cell-based therapies and pharmacological treatments.


Subject(s)
Corneal Injuries , Corneal Opacity , Limbus Corneae , Adult , Humans , Cicatrix , Anti-Inflammatory Agents , Inflammation
2.
Int J Mol Sci ; 23(21)2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36362184

ABSTRACT

In addition to their therapeutic potential in regenerative medicine, human corneal stromal stem cells (CSSCs) could serve as a powerful tool for drug discovery and development. Variations from different donors, their isolation method, and their limited life span in culture hinder the utility of primary human CSSCs. To address these limitations, this study aims to establish and characterize immortalized CSSC lines (imCSSC) generated from primary human CSSCs. Primary CSSCs (pCSSC), isolated from human adult corneoscleral tissue, were transduced with ectopic expression of hTERT, c-MYC, or the large T antigen of the Simian virus 40 (SV40T) to generate imCSSC. Cellular morphology, proliferation capacity, and expression of CSSCs specific surface markers were investigated in all cell lines, including TNFAIP6 gene expression levels in vitro, a known biomarker of in vivo anti-inflammatory efficacy. SV40T-overexpressing imCSSC successfully extended the lifespan of pCSSC while retaining a similar morphology, proliferative capacity, multilineage differentiation potential, and anti-inflammatory properties. The current study serves as a proof-of-concept that immortalization of CSSCs could enable a large-scale source of CSSC for use in regenerative medicine.


Subject(s)
Corneal Stroma , Stromal Cells , Adult , Humans , Cell Differentiation/physiology , Cell Line , Stem Cells
3.
Prog Retin Eye Res ; 87: 101011, 2022 03.
Article in English | MEDLINE | ID: mdl-34530154

ABSTRACT

The cornea is the outmost layer of the eye, unique in its transparency and strength. The cornea not only transmits the light essential for vision, also refracts light, giving focus to images. Each of the three layers of the cornea has properties essential for the function of vision. Although the epithelium can often recover from injury quickly by cell division, loss of limbal stem cells can cause severe corneal surface abnormalities leading to corneal blindness. Disruption of the stromal extracellular matrix and loss of cells determining this structure, the keratocytes, leads to corneal opacity. Corneal endothelium is the inner part of the cornea without self-renewal capacity. It is very important to maintain corneal dehydration and transparency. Permanent damage to the corneal stroma or endothelium can be effectively treated by corneal transplantation; however, there are drawbacks to this procedure, including a shortage of donors, the need for continuing treatment to prevent rejection, and limits to the survival of the graft, averaging 10-20 years. There exists a need for new strategies to promote regeneration of the stromal structure and restore vision. This review highlights critical contributions in regenerative medicine with the aim of corneal reconstruction after injury or disease. These approaches include corneal stromal stem cells, corneal limbal stem cells, embryonic stem cells, and other adult stem cells, as well as induced pluripotent stem cells. Stem cell-derived trophic factors in the forms of secretomes or exosomes for corneal regeneration are also discussed. Corneal sensory nerve regeneration promoting corneal transparency is discussed. This article provides description of the up-to-date options for corneal regeneration and presents exciting possible avenues for future studies toward clinical applications for corneal regeneration.


Subject(s)
Corneal Diseases , Corneal Transplantation , Adult , Cornea/physiology , Corneal Diseases/surgery , Corneal Stroma , Humans , Stem Cells/metabolism , Tissue Engineering/methods
4.
Eye Vis (Lond) ; 7(1): 52, 2020 Nov 03.
Article in English | MEDLINE | ID: mdl-33292650

ABSTRACT

BACKGROUND: Corneal stromal stem cells (CSSC) reduce corneal inflammation, prevent fibrotic scarring, and regenerate transparent stromal tissue in injured corneas. These effects rely on factors produced by CSSC to block the fibrotic gene expression. This study investigated the mechanism of the scar-free regeneration effect. METHODS: Primary human CSSC (hCSSC) from donor corneal rims were cultivated to passage 3 and co-cultured with mouse macrophage RAW264.7 cells induced to M1 pro-inflammatory phenotype by treatment with interferon-γ and lipopolysaccharides, or to M2 anti-inflammatory phenotype by interleukin-4, in a Transwell system. The time-course expression of human transforming growth factor ß3 (hTGFß3) and hTGFß1 were examined by immunofluorescence and qPCR. TGFß3 knockdown for > 70% in hCSSC [hCSSC-TGFß3(si)] was achieved by small interfering RNA transfection. Naïve CSSC and hCSSC-TGFß3(si) were transplanted in a fibrin gel to mouse corneas, respectively, after wounding by stromal ablation. Corneal clarity and the expression of mouse inflammatory and fibrosis genes were examined. RESULTS: hTGFß3 was upregulated by hCSSC when co-cultured with RAW cells under M1 condition. Transplantation of hCSSC to wounded mouse corneas showed significant upregulation of hTGFß3 at days 1 and 3 post-injury, along with the reduced expression of mouse inflammatory genes (CD80, C-X-C motif chemokine ligand 5, lipocalin 2, plasminogen activator urokinase receptor, pro-platelet basic protein, and secreted phosphoprotein 1). By day 14, hCSSC treatment significantly reduced the expression of fibrotic and scar tissue genes (fibronectin, hyaluronan synthase 2, Secreted protein acidic and cysteine rich, tenascin C, collagen 3a1 and α-smooth muscle actin), and the injured corneas remained clear. However, hCSSC-TGFß3(si) lost these anti-inflammatory and anti-scarring functions, and the wounded corneas showed intense scarring. CONCLUSION: This study has demonstrated that the corneal regenerative effect of hCSSC is mediated by TGFß3, inducing a scar-free tissue response.

5.
Exp Eye Res ; 200: 108270, 2020 11.
Article in English | MEDLINE | ID: mdl-32979396

ABSTRACT

Corneal opacities affect vision for millions of individuals worldwide. Fibrotic scar tissues accumulate in reaction to inflammatory responses and remain permanently in corneal stroma, and conventionally correctable only by donor corneal transplantation. Numerous studies have explored innovative approaches to reverse corneal scarring through non-surgical means; however, existing mouse models limit these studies, due to the lack of visibility of scar tissue in mouse corneas with steep curvature. Here, we reported that corneal scarring was modelled using a transgenic mouse line, Tg(Col3a1-EGFP)DJ124Gsat, in which enhanced green fluorescence protein (EGFP) reporter expression was driven by the promoter of collagen 3a1 (COL3a1), a stromal fibrosis gene. Similar to wildtype, Col3a1-EGFP transgenic corneas developed opacities after wounding by alkali burn and mechanical ablation, respectively, as examined under stereomicroscopy and Spectral Domain optical coherent tomography. The time course induction of EGFP was aligned with Col3a1 upregulation and matched with the elevated expression of other fibrosis genes (α-smooth muscle actin, fibronectin and tenascin C). Measured by flow cytometry and enzyme-linked immunosorbent assay, increased number of EGFP expressing cells and fluorescent intensities were correlated to corneal thickening and scar volume. After treatment with human corneal stromal stem cells or their exosomes, EGFP expression was downregulated together with the reduction of scar volume and fibrosis gene expression. These results have demonstrated that the transgenic mouse line, Tg(Col3a1-EGFP)DJ124Gsat, can be a valuable tool for the detection of corneal fibrosis and scarring in vivo, and will be useful in monitoring the changes of corneal fibrosis over time.


Subject(s)
Cicatrix/diagnosis , Collagen Type III/genetics , Corneal Injuries/diagnosis , Corneal Stroma/pathology , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Animals , Cicatrix/genetics , Cicatrix/metabolism , Collagen Type III/biosynthesis , Corneal Injuries/genetics , Corneal Injuries/metabolism , Corneal Stroma/metabolism , Enzyme-Linked Immunosorbent Assay , Green Fluorescent Proteins/biosynthesis , Humans , Mice , Mice, Transgenic , RNA/genetics
6.
Exp Eye Res ; 196: 108062, 2020 07.
Article in English | MEDLINE | ID: mdl-32442558

ABSTRACT

The study of corneal stromal keratocytes is motivated by its strong association with corneal health and visual function. They play a dominant role in the maintenance of corneal homeostasis and transparency through the production of collagens, proteoglycans and corneal crystallins. Trauma-induced apoptosis of keratocytes and replacement by fibroblasts and myofibroblasts disrupt the stromal matrix organization, resulting in corneal haze formation and vision loss. It is, therefore, important to understand the biology and behaviours of keratocytes and the associated stromal cell types (like fibroblasts, myofibroblasts, stromal stem cells) in wound healing, corneal pathologies (including keratoconus, keratitis, endothelial disorders) as well as different ophthalmic situations (such as collagen crosslinking/photodynamic treatment, keratoplasty and refractive surgery, and topical medications). The recent development of ex vivo propagation of keratocytes and stromal stem cells, and their translational applications, either via stromal injection or incorporated in bioscaffold, have been shown to restore the corneal transparency and regenerate native stromal tissue in animal models of corneal haze and other disorders.


Subject(s)
Corneal Keratocytes/physiology , Corneal Stroma/cytology , Cells, Cultured , Cornea/physiology , Humans , Stem Cells/physiology
7.
Invest Ophthalmol Vis Sci ; 60(8): 3013-3023, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31310658

ABSTRACT

Purpose: Mesenchymal stem cells (MSCs) have been extensively studied for their capacity to enhance wound healing and represent a promising research field for generating cell therapies for corneal scars. In the present study, we investigated MSCs from different tissues and their potential to differentiate toward corneal keratocytes. Methods: Adipose-derived stem cells, bone marrow MSCs, umbilical cord stem cells, and corneal stromal stem cells (CSSCs) were characterized by their expression of surface markers CD105, CD90, and CD73, and their multilineage differentiation capacity into adipocytes, osteoblasts, and chondrocytes. MSCs were also evaluated for their potential to differentiate toward keratocytes, and for upregulation of the anti-inflammatory protein TNFα-stimulated gene-6 (TNFAIP6) after simulation by IFN-γ and TNF-α. Results: Keratocyte lineage induction was achieved in all MSCs as indicated by the upregulated expression of keratocyte markers, including keratocan, lumican, and carbohydrate sulfotransferase. TNFAIP6 response to inflammatory stimulation was observed only in CSSCs; increasing by 3-fold compared with the control (P < 0.05). Conclusions: Based on our findings, CSSCs appeared to have the greatest differentiation potential toward the keratocyte lineage and the greatest anti-inflammatory properties in vitro.


Subject(s)
Cell Adhesion Molecules/genetics , Corneal Keratocytes/cytology , Gene Expression Regulation , Mesenchymal Stem Cells/cytology , Cell Adhesion Molecules/biosynthesis , Cell Differentiation , Cells, Cultured , Corneal Keratocytes/metabolism , Flow Cytometry , Humans , Mesenchymal Stem Cells/metabolism , RNA/genetics , Tumor Necrosis Factor-alpha
8.
Stem Cells Transl Med ; 8(11): 1192-1201, 2019 11.
Article in English | MEDLINE | ID: mdl-31290598

ABSTRACT

Mesenchymal stem cells from corneal stromal stem cells (CSSC) prevent fibrotic scarring and stimulate regeneration of transparent stromal tissue after corneal wounding in mice. These effects rely on the ability of CSSC to block neutrophil infiltration into the damaged cornea. The current study investigated the hypothesis that tissue regeneration by CSSC is mediated by secreted extracellular vesicles (EVs). CSSC produced EVs 130-150 nm in diameter with surface proteins that include CD63, CD81, and CD9. EVs from CSSC reduced visual scarring in murine corneal wounds as effectively as did live cells, but EVs from human embryonic kidney (HEK)293T cells had no regenerative properties. CSSC EV treatment of wounds decreased expression of fibrotic genes Col3a1 and Acta2, blocked neutrophil infiltration, and restored normal tissue morphology. CSSC EVs labeled with carboxyfluorescein succinimidyl ester dye, rapidly fused with corneal epithelial and stromal cells in culture, transferring microRNA (miRNA) to the target cells. Knockdown of mRNA for Alix, a component of the endosomal sorting complex required for transport, using siRNA, resulted in an 85% reduction of miRNA in the secreted EVs. The EVs with reduced miRNA were ineffective at blocking corneal scarring. Furthermore, CSSC with reduced Alix expression also lost their regenerative function, suggesting EVs as an obligate component in the delivery of miRNA. The results of these studies support an essential role for extracellular vesicles in the process by which CSSC cells block scarring and initiate regeneration of transparent corneal tissue after wounding. EVs appear to serve as a delivery vehicle for miRNA, which affects the regenerative action. Stem Cells Translational Medicine 2019;8:1192-1201.


Subject(s)
Corneal Diseases/therapy , Extracellular Vesicles/transplantation , Fibrosis/therapy , Inflammation/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , MicroRNAs/administration & dosage , Animals , Female , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Wound Healing
9.
Stem Cells Transl Med ; 7(6): 487-494, 2018 06.
Article in English | MEDLINE | ID: mdl-29654654

ABSTRACT

Stem cells from human corneal stroma (CSSC) suppress corneal stromal scarring in a mouse wound-healing model and promote regeneration of native transparent tissue (PMID:25504883). This study investigated efficacy of compressed collagen gel (CCG) as a vehicle to deliver CSSC for corneal therapy. CSSC isolated from limbal stroma of human donor corneas were embedded in soluble rat-tendon collagen, gelled at 37°C, and partially dehydrated to a thickness of 100 µm by passive absorption. The CCG disks were dimensionally stable, easy to handle, and could be adhered securely to de-epithelialized mouse cornea with fibrin-based adhesive. CSSC in CCG maintained >80% viability for >1 week in culture media and could be cryopreserved in 20% fetal bovine serum-10%DMSO in liquid nitrogen. CCG containing as few as 500 CSSC effectively prevented visible scarring and suppressed expression of fibrotic Col3a1 mRNA. CSSC in CCG were more effective at blocking scarring on a per-cell basis than CSSC delivered directly in a fibrin gel as previously described. Collagen-embedded cells retained the ability to suppress corneal scarring after conventional cryopreservation. This study demonstrates use of a common biomaterial that can facilitate storage and handling of stem cells in a manner that may provide off-the-shelf delivery of stem cells as a therapy for corneal scarring. Stem Cells Translational Medicine 2018;7:487-494.


Subject(s)
Cicatrix/therapy , Collagen/chemistry , Stem Cell Transplantation , Stem Cells/cytology , Animals , Cell Survival/drug effects , Cell- and Tissue-Based Therapy , Cornea/cytology , Cornea/pathology , Cryopreservation , Disease Models, Animal , Female , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Mice , Stem Cells/drug effects , Stem Cells/metabolism , Tissue Engineering/methods , Tissue Scaffolds
10.
J Tissue Eng Regen Med ; 12(1): 59-69, 2018 01.
Article in English | MEDLINE | ID: mdl-27863068

ABSTRACT

Blinding corneal scarring is predominately treated with allogeneic graft tissue; however, there is a worldwide shortage of donor tissue leaving millions in need of therapy. Human corneal stromal stem cells (CSSC) have been shown produce corneal tissue when cultured on nanofibre scaffolding, but this tissue cannot be readily separated from the scaffold. In this study, scaffold-free tissue engineering methods were used to generate biomimetic corneal stromal tissue constructs that can be transplanted in vivo without introducing the additional variables associated with exogenous scaffolding. CSSC were cultured on substrates with aligned microgrooves, which directed parallel cell alignment and matrix organization, similar to the organization of native corneal stromal lamella. CSSC produced sufficient matrix to allow manual separation of a tissue sheet from the grooved substrate. These constructs were cellular and collagenous tissue sheets, approximately 4 µm thick and contained extracellular matrix molecules typical of corneal tissue including collagen types I and V and keratocan. Similar to the native corneal stroma, the engineered corneal tissues contained long parallel collagen fibrils with uniform diameter. After being transplanted into mouse corneal stromal pockets, the engineered corneal stromal tissues became transparent, and the human CSSCs continued to express human corneal stromal matrix molecules. Both in vitro and in vivo, these scaffold-free engineered constructs emulated stromal lamellae of native corneal stromal tissues. Scaffold-free engineered corneal stromal constructs represent a novel, potentially autologous, cell-generated, biomaterial with the potential for treating corneal blindness. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Corneal Stroma/physiology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Corneal Stroma/ultrastructure , Extracellular Matrix/metabolism , Humans , Implants, Experimental , Mice , Mice, Inbred C57BL , Stem Cells/cytology
11.
PLoS One ; 12(3): e0171712, 2017.
Article in English | MEDLINE | ID: mdl-28257425

ABSTRACT

Corneal scarring limits vision for millions of individuals worldwide. Corneal transplantation (keratoplasty) is the standard of care for corneal opacity; however, it bears the risk of graft rejection and infection and is not universally available. Stem cell therapy holds promise as an alternative to keratoplasty. Stem cells from human corneal stroma (CSSC) induce regeneration of transparent corneal tissue in a mouse wound-healing model. In this study we investigated the mechanism by which CSSC prevent deposition of fibrotic tissue. Infiltration by CD11b+/Ly6G+ neutrophils and myeloperoxidase expression were increased in corneas 24 hr after corneal wounding but were reduced in CSSC-treated wounds. Secretion of TSG-6, a protein known to regulate neutrophil migration, was up-regulated in CSSC in response to TNFα and as CSSC differentiate to keratocytes. In vivo, wounded mouse corneas treated with CSSC contained human TSG-6. Inhibition of neutrophil infiltration into cornea by CSSC was reversed when TSG-6 expression was knocked down using siRNA. Silencing of TSG-6 expression in CSSC reduced their ability to block scarring and the expression of mRNA for fibrosis-associated proteins collagen III, tenascin C, and smooth muscle actin in wounded corneas. Neutropenic mice exhibited a significant reduction in corneal scarring and fibrotic mRNA expression 2 weeks after wounding. These results support the conclusion that neutrophil infiltration is an essential event in the fibrotic response to corneal damage and that prevention of scarring by CSSC is mediated by secretion of TSG-6 by these cells.


Subject(s)
Corneal Injuries/therapy , Corneal Keratocytes/transplantation , Corneal Stroma/transplantation , Stem Cell Transplantation , Animals , Cell Adhesion Molecules/genetics , Cornea/metabolism , Cornea/physiopathology , Corneal Injuries/physiopathology , Corneal Stroma/physiopathology , Corneal Transplantation , Graft Rejection/physiopathology , Humans , Mice , Neutrophil Infiltration/genetics , Regeneration/genetics , Wound Healing
12.
Sci Rep ; 6: 30987, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27527833

ABSTRACT

Given the increasing emergence of antimicrobial resistant microbes and the near absent development of new antibiotic classes, innovative new therapeutic approaches to address this global problem are necessary. The use of predatory bacteria, bacteria that prey upon other bacteria, is gaining interest as an "out of the box" therapeutic treatment for multidrug resistant pathogenic bacterial infections. Before a new antimicrobial agent is used to treat infections, it must be tested for safety. The goal of this study was to test the tolerability of bacteria on the ocular surface using in vitro and in vivo models. Predatory bacteria Bdellovibrio bacteriovorus and Micavibrio aeruginosavorus were found to be non-toxic to human corneal stromal keratocytes in vitro; however, they did induce production of the proinflammatory chemokine IL-8 but not IL-1ß. Predatory bacteria did not induce inflammation on the ocular surface of rabbit eyes, with and without corneal epithelial abrasions. Unlike a standard of care antibiotic vancomycin, predatory bacteria did not inhibit corneal epithelial wound healing or increase clinical inflammatory signs in vivo. Together these data support the safety of predatory bacteria on the ocular surface, but future studies are warranted regarding the use predatory bacteria in deeper tissues of the eye.


Subject(s)
Alphaproteobacteria/growth & development , Antibiosis , Bdellovibrio bacteriovorus/growth & development , Eye/immunology , Inflammation/immunology , Wound Healing/physiology , Animals , Eye/microbiology , Female , Humans , Inflammation/microbiology , Rabbits
13.
J Cataract Refract Surg ; 42(2): 302-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27026456

ABSTRACT

PURPOSE: To use a well-established organ culture model to investigate the effects of corneal stromal stem cells on the optical and biomechanical properties of corneal wounds after laser in situ keratomileusis (LASIK)-like flap creation. SETTING: School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, United Kingdom. DESIGN: Experimental study. METHODS: The LASIK-like flaps were produced in sheep corneas. The flap beds were treated with corneal stromal stem cells and were then replaced and allowed to heal for different periods of up to 3 weeks in organ culture. The optical transmission of the cornea, the force required to detach the flap, and the presence of myofibroblasts near the flap bed were measured. RESULTS: Corneal stromal stem cell-treated flap beds were statistically significantly more transparent after 3 weeks in culture than the untreated controls. At 3 weeks, the mean force necessary to detach the flap was more than twice the force required for the respective control samples. Concurrently, there were 44% activated cells immediately below the flap margin of the controls compared with 29% in the same region of the corneal stromal stem cell-treated flaps. CONCLUSIONS: In this system, the presence of corneal stromal stem cells at the wound margin significantly increased the adherence of LASIK-like flaps while maintaining corneal transparency. It is postulated that this is achieved by the deposition of extracellular connective tissue similar to that found in the normal cornea and by the paucity of activated keratocytes (myofibroblasts), which are known to scatter a significant amount of the incident light. FINANCIAL DISCLOSURE: No author has a financial or proprietary interest in any material or method mentioned.


Subject(s)
Cornea/surgery , Corneal Stroma/cytology , Keratomileusis, Laser In Situ , Lasers, Excimer , Stem Cell Transplantation , Stem Cells/physiology , Wound Healing/physiology , Actins/metabolism , Animals , Cornea/physiology , Corneal Pachymetry , Humans , Microscopy, Fluorescence , Organ Culture Techniques , Sheep , Surgical Flaps/physiology , Tissue Adhesions
14.
Ocul Surf ; 14(2): 113-20, 2016 04.
Article in English | MEDLINE | ID: mdl-26804252

ABSTRACT

The corneal stroma contains a population of mesenchymal cells subjacent to the limbal basement membrane with characteristics of adult stem cells. These 'niche cells' support limbal epithelial stem cell viability. In culture by themselves, the niche cells display a phenotype typical of mesenchymal stem cells. These stromal stem cells exhibit a potential to differentiate to multiple cell types, including keratocytes, thus providing an abundant source of these rare cells for experimental and bioengineering applications. Stromal stem cells have also shown the ability to remodel pathological stromal tissue, suppressing inflammation and restoring transparency. Because stromal stem cells can be obtained by biopsy, they offer a potential for autologous stem cell treatment for stromal opacities. This review provides an overview of the status of work on this interesting cell population.


Subject(s)
Stem Cells , Corneal Stroma , Epithelium, Corneal , Humans , Limbus Corneae
15.
Stem Cells Transl Med ; 4(3): 276-85, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25713466

ABSTRACT

Corneal blindness afflicts millions of individuals worldwide and is currently treated by grafting with cadaveric tissues; however, there are worldwide donor tissue shortages, and many allogeneic grafts are eventually rejected. Autologous stem cells present a prospect for personalized regenerative medicine and an alternative to cadaveric tissue grafts. Dental pulp contains a population of adult stem cells and, similar to corneal stroma, develops embryonically from the cranial neural crest. We report that adult dental pulp cells (DPCs) isolated from third molars have the capability to differentiate into keratocytes, cells of the corneal stoma. After inducing differentiation in vitro, DPCs expressed molecules characteristic of keratocytes, keratocan, and keratan sulfate proteoglycans at both the gene and the protein levels. DPCs cultured on aligned nanofiber substrates generated tissue-engineered, corneal stromal-like constructs, recapitulating the tightly packed, aligned, parallel fibrillar collagen of native stromal tissue. After injection in vivo into mouse corneal stroma, human DPCs produced corneal stromal extracellular matrix containing human type I collagen and keratocan and did not affect corneal transparency or induce immunological rejection. These findings demonstrate a potential for the clinical application of DPCs in cellular or tissue engineering therapies for corneal stromal blindness.


Subject(s)
Blindness/therapy , Cell Differentiation , Cornea/metabolism , Corneal Diseases/therapy , Dental Pulp/cytology , Stem Cell Transplantation , Stem Cells/cytology , Adult , Animals , Blindness/metabolism , Corneal Diseases/metabolism , Dental Pulp/metabolism , Heterografts , Humans , Mice , Stem Cells/metabolism
16.
Sci Transl Med ; 6(266): 266ra172, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25504883

ABSTRACT

Conventional allograft therapy for corneal scarring is widespread and successful, but donor tissue is not universally available, and some grafts fail owing to rejection and complications such as endothelial failure. We investigated direct treatment of corneal scarring using autologous stem cells, a therapy that, if successful, could reduce the need for corneal grafts. Mesenchymal cells were expanded from small superficial, clinically replicable limbal biopsies of human cadaveric corneo-scleral rims. Limbal biopsy-derived stromal cells (LBSCs) expanded rapidly in media containing human serum, were highly clonogenic, and could generate spheres expressing stem cell genes (ABCG2, Nestin, NGFR, Oct4, PAX6, and Sox2). Human LBSCs differentiated into keratocytes expressing characteristic marker genes (ALDH3A1, AQP1, KERA, and PTGDS) and organized a thick lamellar stroma-like tissue containing aligned collagen and keratan sulfate proteoglycans when cultured on aligned nanofiber substrata. When engrafted into mouse corneal wounds, LBSCs prevented formation of light-scattering scar tissue containing fibrotic matrix components. The presence of LBSCs induced regeneration of ablated stroma with tissue exhibiting lamellar structure and collagen organization indistinguishable from that of native tissue. Because the limbus can be easily biopsied from either eye of an affected individual and LBSCs capable of corneal stromal remodeling can be expanded under xeno-free autologous conditions, these cells present a potential for autologous stem cell-based treatment of corneal stromal blindness.


Subject(s)
Cornea/pathology , Corneal Transplantation/methods , Stem Cell Transplantation , Stem Cells/cytology , Stromal Cells/cytology , Adult , Animals , Biopsy , Cell Differentiation , Cicatrix/prevention & control , Collagen/chemistry , Cornea/metabolism , Culture Media/chemistry , Female , Fibroblasts/cytology , Gene Expression Regulation , Humans , Keratan Sulfate/chemistry , Keratinocytes/cytology , Mice , Mice, Inbred C57BL , Middle Aged , Proteoglycans/chemistry
17.
Biomaterials ; 35(12): 3744-55, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24503156

ABSTRACT

Emulating corneal stromal tissue is believed to be the most challenging step in bioengineering an artificial human cornea because of the difficulty in reproducing its highly ordered microstructure, the key to the robust biomechanical properties and optical transparency of this tissue. We conducted a comparative study to assess the feasibility of human corneal stromal stem cells (hCSSCs) and human corneal fibroblasts (hCFs) in the generation of human corneal stromal tissue on groove-patterned silk substrates. In serum-free keratocyte differentiation medium, hCSSCs successfully differentiated into keratocytes secreting multilayered lamellae with orthogonally-oriented collagen fibrils, in a pattern mimicking human corneal stromal tissue. The constructs were 90-100 µm thick, containing abundant cornea-specific extracellular matrix (ECM) components, including keratan sulfate, lumican, and keratocan. In contrast, hCFs tended to differentiate into myofibroblasts that deposited less organized collagen in a pattern resembling that of corneal scar tissue. RGD surface coupling coupling was an essential factor in enhancing cell attachment, orientation, proliferation, differentiation and ECM deposition on the silk substratum. These results demonstrated that an approach of combining hCSSCs with an RGD surface-coupled patterned silk film offers a powerful tool to develop highly ordered collagen fibril-based constructs for corneal regeneration and corneal stromal tissue repair.


Subject(s)
Cornea/cytology , Silk , Stromal Cells/cytology , Tissue Scaffolds , Cell Differentiation , Cells, Cultured , Cornea/metabolism , Culture Media, Serum-Free , Extracellular Matrix Proteins/metabolism , Gene Expression , Humans , Keratinocytes/cytology , Stromal Cells/metabolism
18.
PLoS One ; 9(1): e86260, 2014.
Article in English | MEDLINE | ID: mdl-24465995

ABSTRACT

Human corneal fibroblasts (HCF) and corneal stromal stem cells (CSSC) each secrete and organize a thick stroma-like extracellular matrix in response to different substrata, but neither cell type organizes matrix on tissue-culture polystyrene. This study compared cell differentiation and extracellular matrix secreted by these two cell types when they were cultured on identical substrata, polycarbonate Transwell filters. After 4 weeks in culture, both cell types upregulated expression of genes marking differentiated keratocytes (KERA, CHST6, AQP1, B3GNT7). Absolute expression levels of these genes and secretion of keratan sulfate proteoglycans were significantly greater in CSSC than HCF. Both cultures produced extensive extracellular matrix of aligned collagen fibrils types I and V, exhibiting cornea-like lamellar structure. Unlike HCF, CSSC produced little matrix in the presence of serum. Construct thickness and collagen organization was enhanced by TGF-ß3. Scanning electron microscopic examination of the polycarbonate membrane revealed shallow parallel grooves with spacing of 200-300 nm, similar to the topography of aligned nanofiber substratum which we previously showed to induce matrix organization by CSSC. These results demonstrate that both corneal fibroblasts and stromal stem cells respond to a specific pattern of topographical cues by secreting highly organized extracellular matrix typical of corneal stroma. The data also suggest that the potential for matrix secretion and organization may not be directly related to the expression of molecular markers used to identify differentiated keratocytes.


Subject(s)
Collagen/metabolism , Cornea/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Stem Cells/metabolism , Cell Differentiation/physiology , Cells, Cultured , Cornea/physiology , Corneal Keratocytes/metabolism , Corneal Keratocytes/physiology , Corneal Stroma/metabolism , Corneal Stroma/physiology , Corneal Topography/methods , Cues , Extracellular Matrix/physiology , Fibroblasts/physiology , Humans , Keratan Sulfate/metabolism , Stem Cells/physiology , Transforming Growth Factor beta3/metabolism
19.
PLoS One ; 8(2): e56831, 2013.
Article in English | MEDLINE | ID: mdl-23437251

ABSTRACT

Corneal transparency depends on a unique extracellular matrix secreted by stromal keratocytes, mesenchymal cells of neural crest lineage. Derivation of keratocytes from human embryonic stem (hES) cells could elucidate the keratocyte developmental pathway and open a potential for cell-based therapy for corneal blindness. This study seeks to identify conditions inducing differentiation of pluripotent hES cells to the keratocyte lineage. Neural differentiation of hES cell line WA01(H1) was induced by co-culture with mouse PA6 fibroblasts. After 6 days of co-culture, hES cells expressing cell-surface NGFR protein (CD271, p75NTR) were isolated by immunoaffinity adsorption, and cultured as a monolayer for one week. Keratocyte phenotype was induced by substratum-independent pellet culture in serum-free medium containing ascorbate. Gene expression, examined by quantitative RT-PCR, found hES cells co-cultured with PA6 cells for 6 days to upregulate expression of neural crest genes including NGFR, SNAI1, NTRK3, SOX9, and MSX1. Isolated NGFR-expressing cells were free of PA6 feeder cells. After expansion as a monolayer, mRNAs typifying adult stromal stem cells were detected, including BMI1, KIT, NES, NOTCH1, and SIX2. When these cells were cultured as substratum-free pellets keratocyte markers AQP1, B3GNT7, PTDGS, and ALDH3A1 were upregulated. mRNA for keratocan (KERA), a cornea-specific proteoglycan, was upregulated more than 10,000 fold. Culture medium from pellets contained high molecular weight keratocan modified with keratan sulfate, a unique molecular component of corneal stroma. These results show hES cells can be induced to differentiate into keratocytes in vitro. Pluripotent stem cells, therefore, may provide a renewable source of material for development of treatment of corneal stromal opacities.


Subject(s)
Cell Differentiation , Corneal Keratocytes/cytology , Embryonic Stem Cells/cytology , Animals , Cell Line , Coculture Techniques , Corneal Keratocytes/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Humans , Immunophenotyping , Mice , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Organ Specificity/genetics , Phenotype , Proteoglycans/metabolism , Receptor, Nerve Growth Factor/metabolism , Stromal Cells/metabolism , Sulfotransferases/genetics , Sulfotransferases/metabolism , Carbohydrate Sulfotransferases
20.
Invest Ophthalmol Vis Sci ; 53(3): 1566-75, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22297497

ABSTRACT

PURPOSE: To isolate and characterize stem cells from human trabecular meshwork (TM) and to investigate the potential of these stem cells to differentiate into TM cells. METHODS: Human trabecular meshwork stem cells (TMSCs) were isolated as side population cells by fluorescence-activated cell sorting or isolated by clonal cultures. Passaged TMSCs were compared with primary TM cells by immunostaining and quantitative RT-PCR. TMSC purity was assessed by flow cytometry and TMSC multipotency was examined by induction of neural cells, adipocytes, keratocytes, or TM cells. Differential gene expression was detected by quantitative RT-PCR, immunostaining, and immunoblotting. TM cell function was evaluated by phagocytic assay using inactivated Staphylococcus aureus bioparticles. RESULTS: Side population and clonal isolated cells expressed stem cell markers ABCG2, Notch1, OCT-3/4, AnkG, and MUC1 but not TM markers AQP1, MGP, CHI3L1, or TIMP3. Passaged TMSCs are a homogeneous population with >95% cells positive to CD73, CD90, CD166, or Bmi1. TMSCs exhibited multipotent ability of differentiation into a variety of cell types with expression of neural markers neurofilament, ß-tubulin III, GFAP; or keratocyte-specific markers keratan sulfate and keratocan; or adipocyte markers ap2 and leptin. TMSC readily differentiated into TM cells with phagocytic function and expression of TM markers AQP1, CHI3L1, and TIMP3. CONCLUSIONS: TMSCs, isolated as side population or as clones, express specific stem cell markers, are homogeneous and multipotent, with the ability to differentiate into phagocytic TM cells. These cells offer a potential for development of a novel stem cell-based therapy for glaucoma.


Subject(s)
Cell Differentiation , Multipotent Stem Cells/cytology , Phagocytes/cytology , Trabecular Meshwork/cytology , Biomarkers/metabolism , Cells, Cultured , Corneal Keratocytes/cytology , Flow Cytometry , Humans , Immunoblotting , Multipotent Stem Cells/metabolism , Phagocytosis/physiology , Reverse Transcriptase Polymerase Chain Reaction , Trabecular Meshwork/metabolism , Trabecular Meshwork/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...